Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Sep 8.
Published in final edited form as: Biochim Biophys Acta. 2008 Oct 11;1792(1):61–67. doi: 10.1016/j.bbadis.2008.09.016

GALNT3, a gene associated with hyperphosphatemic familial tumoral calcinosis, is transcriptionally regulated by extracellular phosphate and modulates matrix metalloproteinase activity

Ilana Chefetz a,b, Kimitoshi Kohno c, Hiroto Izumi c, Jouni Uitto d, Gabriele Richard d,e, Eli Sprecher a,b,*
PMCID: PMC3169302  NIHMSID: NIHMS317678  PMID: 18976705

Abstract

GALNT3 encodes UDP-N-acetyl-alpha-D-galactosamine: polypeptide N-acetylgalactosaminyl-transferarase 3 (ppGalNacT3), a glycosyltransferase which has been suggested to prevent proteolysis of FGF23, a potent phosphaturic protein. Accordingly, loss-of-function mutations in GALNT3 cause hyperphosphatemic familial tumoral calcinosis (HFTC), a rare autosomal recessive disorder manifesting with increased kidney reabsorption of phosphate, resulting in severe hyperphosphatemia and widespread ectopic calcifications. Although these findings definitely attribute a role to ppGalNacT3 in the regulation of phosphate homeostasis, little is currently known about the factors regulating GALNT3 expression. In addition, the effect of decreased GALNT3 expression in peripheral tissues has not been explored so far. In the present study, we demonstrate that GALNT3 expression is under the regulation of a number of factors known to be associated with phosphate homeostasis, including inorganic phosphate itself, calcium and 1,25-dihydroxyvitamin D3. In addition, we show that decreased GALNT3 expression in human skin fibroblasts leads to increased expression of FGF7 and of matrix metalloproteinases, which have been previously implicated in the pathogenesis of ectopic calcification. Thus, the present data suggest that ppGalNacT3 may play a role in peripheral tissues of potential relevance to the pathogenesis of disorders of phosphate metabolism.

Keywords: Calcinosis, Phosphate, Calcification

1. Introduction

Inorganic phosphate (Pi) is an element of essential importance for skeletal mineralization, mineral homeostasis, and a wide range of cellular processes involving signaling and synthesis of energy-generating compounds. Pi dietary intake, renal and intestinal absorption and exchange with intracellular and bone storage pools, are tightly regulated to maintain its serum level within a narrow range [1]. Elevated Pi levels have been linked to increased mortality in patients with end-stage or even early stage chronic kidney disease [2,3]. Moreover, serum Pi levels in the upper normal range were found to be associated with a significant increase in mortality among individuals with normal renal function [4].

Although hyperphosphatemia is associated with many common acquired conditions[5], only a few inherited (monogenic) disorders manifesting with increased serum Pi levels are known [6]. Hyperphosphatemic familial tumoral calcinosis (HFTC; MIM211900) is a rare autosomal recessive condition characterized by the progressive deposition of basic calcium phosphate crystals in peri-articular spaces and soft tissues. HFTC is associated with marked hyperphosphatemia in the face of inappropriately normal parathyroid (PTH) and normal or slightly elevated levels of 1,25-dihydroxyvitamin D (1,25(OH)3D ) [7]. HFTC may result from mutations in three different genes, encoding proteins which are part of a common metabolic pathway. The first mutations associated with HFTC were initially reported in GALNT3 [815], which encodes UDP-N-acetyl-alpha-D-galactosamine: polypeptide N-acetylgalactosaminyl-transferarase 3 (ppGalNacT3), a glycosyltransferase that initiates mucin-type O-glycosylation [16]. Loss of function mutations in the fibroblast growth factor 23 (FGF23) gene were subsequently found to also cause HFTC [1720]. FGF23 codes for a circulating molecule that promotes renal phosphate excretion by decreasing phosphate re-absorption in the proximal renal tubule. In addition, FGF23 further contributes to lowering circulating Pi levels by down-regulating the biosynthesis and increasing the catabolism of 1,25 (OH)3D [21,22], resulting in decreased 1,25(OH)3D levels. ppGalNacT3 was shown in vitro to be essential for FGF23 processing and secretion; moreover, decreased ppGalNacT3 activity was found to result in decreased levels of active FGF23 in the circulation [2325]. Recently, a mutation in a third gene, KL, was found to cause a phenotype very much resembling HFTC [26,27]. KL encodes Klotho, which functions as a co-receptor for FGF23 [28].

Although it is clear that ppGalNacT3, FGF23 or Klotho deficiency affects Pi homeostasis systemically through dysregulation of phosphate reabsorption in the kidney (and possibly intestinal mucosa [29]), little is currently known about non-systemic, kidney-independent, features of ppGalNacT3 regulation and metabolic effects. Interestingly, ectopic calcification in HFTC is prominently (although not exclusively) observed in cutaneous and subcutaneous tissues [7]. To investigate these aspects of ppGalNacT3 physiological roles, we used in vitro systems and identified Pi as a major regulator of GALNT3 gene expression. In addition, we found out that as a consequence of decreased ppGalNacT3 expression, fibroblasts release increased amounts of matrix metalloproteinases (MMPs), which have been previously implicated in the pathogenesis of ectopic calcification [3033]. These data highlight the possible role of ppGalNacT3 in peripheral tissues.

2. Materials and methods

2.1. Materials

Parathyroid Hormone Fragment 1-34 (PTH), 1α,25 Dihydroxyvitamin D3, calcium chloride, and sodium phosphate dibasic dehydrate were obtained from Sigma(St Louis, USA). Phosphonoformic acid (PFA) was purchased from MBL International (Woburn, MA, USA). FGF7 was purchased from Pepro Tech (Rocky Hill, NJ, USA).

2.2. Cell cultures

Primary fibroblast cell cultures were derived from punch biopsies obtained from patients or healthy controls after having received their written and informed consent according to a protocol reviewed and approved by our Institutional Review Board. Primary human fibroblasts immortalized with a hTERT-expressing retroviral vector (hTERT-fibroblasts) [34] were kindly provided by Dr. Sarah Selig. All cells were maintained in DMEM-low glucose medium (Biological Industries, Beith Haemek, Israel) supplemented with 10% fetal calf serum, 1% L-Glutamine and 1% penicillin/streptomycin. Human embryonic kidney cells (HEK 293) were maintained in DMEM/F12 medium with the same supplements.

2.3. Quantitative RT-PCR (qRT-PCR)

For quantitative real-time PCR, cDNA was synthesized from 1 μg of total RNA extracted from cultured fibroblasts using the Reverse-iT first strand synthesis kit (ABgene, Epsom, UK) and random hexamers. cDNA PCR amplification was carried out using the Absolute QPCR SYBR® Green Mix (ABgene) on a Rotor-Gene 3000 multi-filter system (Corbett Research, Sydney, Australia) with primer pairs specific for GALNT3, GALNT1, GALNT4, GALNT6, FGF7, MMP9, MMP8 and ACTB (Table 1). To ensure the specificity of the reaction conditions, at the end of the individual runs, the melting temperature (Tm) of the amplified products was measured to confirm their homogeneity. Cycling conditions were as follows: 95 °C for 10 min; 95 °C for 10 s, 62 °C for 25 s and 72 °C for 15 s for a total of 40 cycles. Each sample was analyzed in triplicate. For quantification, standard curves were obtained using serially diluted cDNA amplified in the same real-time PCR run. Results were normalized to ACTB mRNA levels, and for confirmation to GAPDH mRNA levels. After the quantification procedure, the products were resolved by 2.5% agarose gel electrophoresis to confirm that the reaction had amplified the DNA fragments of expected size.

Table 1.

qRT-PCR oligonucleotide sequences

Sequence Size (bp) Gene
8F-CTCTATGTCTGGATGTTGG 109 GALNT3
9R-TCATGTTGAGCAGAGTATTC
1F-CTTGATTTGGGTACTCTTGG 122 GALNT1
2R-GAGGCTTTTGTACTGGCTCTAG
1F-GTGGAGCTCTTGGTCTCT 187 GALNT4
1R-TCGTTGAGCTGGAGTTTG
1F-ACGAGATGAATGAGGAGC 206 GALNT6
3R-CTGCGTCAGCTCTGAGT
12F-CTTTGGACACGCACGAC 131 MMP9
13R-GGATGTCATAGGTCACGTAGC
1F-GACGCTTCCATTTCTGCT 164 MMP8
2R-CGATCACATTAGTGCCATTC
1F-CAATGACATGACTCCAGAGC 274 FGF7
2R-CAACTGCCACTGTCCTG
3F-CGATCACATTAGTGCCATTC 127 ACTB
4R-AGGTGGACAGCGAGGCCAGGA

2.4. Small interfering RNA (siRNA) transfection

Fibroblasts cells were grown to log phase, trypsinized and maintained in 2 ml of growth medium without antibiotics into 6-well tissue culture plates at 50% confluence for 24 h. Six microliters of lipofectamine (Invitrogen, Carlsbad, CA) were mixed in a total volume of 500 μl of Opti-MEM medium with GALNT3-specific siRNA or scrambled siRNA (20 nM) (Invitrogen) and added to the cells for 6 h, at which time, the transfection medium was replaced with complete growth medium. The GALNT3-specific siRNA duplex used consisted of 5′-rGrGrCrArUrCrArAUrArCrArGrCUUrCrATT-3′ and 5′-UrGrArArGrCUrGUrAUUrGrAUUrGrCrCTT3′.

2.5. Western blotting

Cells were homogenized in lysis buffer (25 mM Hepes, 300 mM NaCl, 1.5 mM MgCl2, 0.2 mM EDTA and protease inhibitors mix including 1 mM PMSF, 1 mg/ml aprotinin and leupeptin; (Sigma, St Louis, MO, USA). Following centrifugation at 10,000 g for 10 min at 4 °C, proteins were electrophoresed through a 10% SDS-PAGE and transferred onto a nitrocellulose membrane (Trans-Blot Bio-Rad, Hercules, CA, USA). After 1 h blocking with 1xTBS (20 mM Tris, 150 mM NaCl) with 5% skim milk and 0.01% Tween 20, blots were incubated with a rabbit polyclonal anti-ppGalNacT3 antibody [35]. The specificity of this antibody has been demonstrated in previous publications [36]. The blots were washed three times with TBS-Tween (20 mM Tris HCl, 4 mM Tris base, 140 mM NaCl, 1 mM EDTA, 0.1% Tween-20). After incubation with secondary HRP-conjugated anti-rabbit antibody (Sigma-Aldrich, St. Louis, MO, USA) and subsequent washings, proteins were detected using the EZ-ECL chemiluminescence detection kit (Biological Industries, Beit Haemek, Israel).

In order to compare the amount of protein in the different samples, the blots were reprobed with mouse monoclonal antibody to β-actin (ABcam, Cambridge, UK) and secondary HRP-conjugated anti-mouse antibody (Sigma-Aldrich, St. Louis, MO, USA). For quantitative determination of signal intensity, membranes were scanned with ImageMaster VDS-Cl (Amersham Pharmacia Biotech) and analyzed by digital densitometry (Tina 2.10 g software).

2.6. FGF7 ELISA

Human FGF7 Eliza Quantikine kit (R and D, Minneapolis, MN, USA) was used to detect FGF7 in the media of cultured primary fibroblasts, according to the manufacturer’s instructions.

2.7. Gelatin zymography

Conditioned medium (CM) was collected from cultured fibroblasts maintained under serum-free conditions for 24 h and concentrated 1:20 using Vivaspin 6 concentrators (Sartorius, Goettingen, Germany). The samples were then mixed with non-reducing SDS gel sample buffer and applied without boiling to a 10% polyacrylamide gel containing 0.1% SDS and 1 mg/ml gelatin. After electrophoresis, the gels were washed in 50 mmol/l Tris-HCl (pH 7.5) containing 0.15 mol/l NaCl, 5 mmol/l CaCl2, 0.02% NaN3, 0.25% Triton X-100 (three changes) at room temperature, and then incubated in the same buffer without Triton X-100 at 37 °C for 20 h. Proteins were stained by Coomassie Brilliant Blue R-250 solution (Bio-Rad, Hercules, CA, USA).

2.8. MMP Profiling

MMP activity in the CMs was determined using Enzolyte MMP fluorometric assay kit (AnaSpec, Inc., San Jose, CA, USA). The CM samples were incubated with MMP-specific peptide substrates following the manufacturer’s instructions. Cleavage of substrates by MMPs removed the quenching effect of QXL520 on 5-carboxyfluorescein, resulting in increased fluorescence with excitation at 490 nm and emission at 535 nm [37]. Quantification of signal emission was performed using FLUOstar galaxy microplate reader (BMG Labtech GmbH, Jena, Germany).

3. Results

3.1. Identification of major regulators of ppGalNacT3/GALNT3 expression in human cells

Although abnormal expression of ppGalNacT3 is clearly associated with marked derangements of Pi metabolism [1,6,7,21,22], it is not known whether Pi itself is involved in the regulation of ppGalNacT3/GALNT3 expression. We therefore assessed the effect of increasing Pi concentrations on ppGalNacT3 expression in human hTERT-fibroblasts in culture. ppGalNacT3 expression was strongly induced in response to Pi (Fig. 1a) up to extracellular concentration of 5 mM, which is known to trigger apoptosis [38]. To determine the molecular basis of Pi-induced ppGalNacT3 up-regulation, we assessed GALNT3 expression by qRT-PCR. Consistent with the induction observed at the protein level, treatment with Pi for 24 h at 3.5 mM up-regulated GALNT3 expression 2.4-fold, whereas lower Pi concentrations had little effect (Fig. 1b). Cells were then cultured in medium with 3.5 mM Pi for different time periods. We observed an initial increase in GALNT3 RNA level 12 h after raising Pi concentrations, reaching maximal induction after 22 h (Fig. 1c).

Fig. 1.

Fig. 1

Effect of Pi on PPGalNacT3/GALNT3 expression in human fibroblasts. (a) hTERT fibroblasts were cultured for 24 h in the presence of increasing concentrations of Pi, and ppGalNacT3 expression was ascertained by Western Blotting (upper panel). Results were quantified by digital densitometry and the expression of ppGalNacT3 was normalized to that of β-actin (Tina 2.10 g software; lower panel); hTERT fibroblasts were cultured for 24 h in the presence of (b) increasing concentrations of Pi or (c) for increasing time periods in the presence of 3.5 mM Pi, and GALNT3 gene expression was quantified by qRT-PCR. Results are expressed as % of expression relative to control cells+ standard deviation (SD).

To determine whether the effect of Pi on GALNT3 expression is cell lineage-specific, we assessed GALNT3 expression in HEK293 cells (a cell line derived from human embryonic kidney tissue) exposed to varying concentrations of Pi. Here again, a Pi dose-dependent increase in ppGalNacT3 protein (Fig. 2a) and GALNT3 RNA (Fig. 2b) levels was observed. GALNT3 gene induction was most most likely dependent upon Pi influx into the cells as the effect of Pi on GALNT3 expression could be blocked by exposing the cells to phosphonoformic acid (PFA), a known inhibitor of Pi transport [39] (Fig. 2c).

Fig. 2.

Fig. 2

Regulation of ppGalNacT3/GALNT3 in HEK293 cells. (a) HEK293 cells were cultured in the presence of 1 mM or 3.5 mM Pi, and ppGalNacT3 expression was assessed by (a) Western blotting and (b) qRT-PCR 24 h later; (c) To assess the effect of Pi transport on GALNT3 induction, we maintained the cells in 3.5 mM Pi in the presence (+) or absence (−) of PFA, a known inhibitor of Pi transport (100% refers to the level of expression of GALNT3 in the presence of 1 mM phosphate); (d) We also measured GALNT3 expression in HEK293 cells cultured with increasing concentrations of calcium and 1,25(OH)3D. Results in (b) and (d) are expressed as % of expression relative to control cells+SD.

We also determine the effects of other molecules previously implicated in the regulation of Pi circulating levels on GALNT3 expression. Although PTH lacked any significant effect (data not shown), both calcium and 1,25(OH)3D had a suppressive dose-dependent effect on GALNT3 gene expression (Fig. 2d).

3.2. GALNT3 down-regulation induces FGF7 expression

As mentioned above, the reason for the preferential accumulation of calcium deposits in the skin and subcutaneous tissues in HFTC [7] is still unclear. Recently, FGF7 (also known as keratinocyte growth factor; KGF) was identified as capable of promoting Pi excretion through the kidney [40]. The major source of FGF7 under physiological conditions are dermal fibroblasts [41]. We therefore postulated that as we found that Pi induces the expression of both GALNT3 (Fig. 1) and FGF7 (Fig. 2S), GALNT3 gene expression may possibly influence the expression of FGF7. To assess this possibility, we used siRNA transfection to down-regulate the expression of GALNT3 in hTERT-fibroblasts (Fig. 1S). Significantly increased FGF7 RNA levels were found in cells transfected with GALNT3-specific siRNA as compared with control siRNA-transfected cells (Fig. 3a). To assess the relevance of these findings to the pathogenesis of HFTC, we also quantified FGF7 gene expression levels in primary fibroblast cell cultures derived from two HFTC patients. These individuals were previously shown to carry a splice site mutation [15], which was found to result in complete absence of the ppGalNAcT3 protein in the skin [42]. GALNT3 gene transcription was decreased by 70% in primary fibroblasts derived from HFTC patients as compared with healthy controls (data not shown). In contrast, FGF7 RNA expression was 3.5 fold higher in patient cells than in control primary fibroblasts (Fig. 3b). Accordingly, HFTC patients’ primary fibroblasts were found to secrete much higher concentrations of FGF7 (80+10 vs 27+5 pg/ml), as compared with control fibroblasts (Fig. 3c).

Fig. 3.

Fig. 3

FGF7 expression in ppGalNacT3-deficient human fibroblasts. (a) hTERT fibroblasts were transfected with GALNT3-specific siRNA (siRNA) or a non-relevant siRNA (control), and FGF7 expression was assessed by qRT-PCR after 3 days. Results are presented as % of expression relative to control cells+SD; (b) Primary human fibroblasts derived from HFTC patients (HFTC) and control individuals (control) were assessed for FGF7 expression by qRT-PCR. Results are given as % of expression relative to control cells+SD; (c) FGF7 secretion in the medium obtained from patient (HFTC) and control (control) primary fibroblasts cultures was determined using a quantitative ELISA assay. Results are expressed in pg/ml+SD.

3.3. GALNT3 down-regulation is associated with augmented MMP activity

Among the various potential downstream paracrine effects of FGF7, whose secretion seems to be triggered by GALNT3 down-regulation, we noted that FGF7 has been shown to induce the activity of several MMPs in different tissues [4346], while MMPs have in turn been previously implicated in the pathogenesis of ectopic calcification [3033]. Thus we first assessed the capacity of FGF7 to trigger MMPs secretion in dermal hTERT-fibroblasts. Gelatin-zymography analysis of CM collected 24 h after exposure to FGF7 revealed an increase in gelatinase activity in response to FGF7 (Fig. 4a).

Fig. 4.

Fig. 4

GALNT3 down-regulation increases MMPs expression and activity. (a) Gelatinase activity in the culture medium was assessed by gel zymography, as described in Materials and methods; (b) Human fibroblasts were transfected with GALNT3-specific siRNA (siRNA) or a non-relevant siRNA (control), and MMP8 and MMP9 gene expression was assessed by qRT-PCR. Results are given as % of expression relative to control cells+SD; (c) The same cells were comparatively assessed for MMP activity using a fluorometric assay as described in Materials and Methods; (d) Gelatinase activity in the culture medium of both types of cells was assessed by gel zymography.

Since GALNT3 down-regulation induces FGF7 secretion (Fig. 3), and FGF7 was found both in this (Fig. 4a) and previous studies [4346] to up-regulate MMP expression, we hypothesized that GALNT3 down-regulation may also affect MMP activity in dermal hTERT-fibroblasts. Indeed, significant up-regulation of MMP9 (1.7-fold) and MMP8 (7.9-fold) mRNA levels was found upon down-regulation of GALNT3 expression by siRNA in human fibroblasts (Fig. 4b) as well as in HFTC primary fibroblasts as compared with control cells (not shown). Of note, increased Pi concentration did not affect the extent of gene induction associated with GALNT3 down-regulation (Fig. 3S) and no change in Pi extracellular concentration was observed 3 and 5 days after siRNA transfection (not shown).

We then assessed human hTERT-fibroblasts transfected with GALNT3-specific siRNA for MMP activity using a panel of MMP-specific fluorescent substrates. We observed a significant elevation in the specific activities of MMP-8 (collagenase), MMP-9/2 (gelatinases) and MMP-3 (but not MMP-1) in cells down-regulated for GALNT3 expression as compared with control fibroblasts (Fig. 4c). Gel zymography analysis demonstrated increased gelatinase activity in CM obtained from GALNT3-down-regulated hTERT-fibroblasts as compared with control cells (Fig. 4d).

4. Discussion

Pi homeostasis has been shown to play an important role in human physiology, determining to a large extent survival in selected groups [5,47,48]. The study of rare inherited disorders often leads to insights into physiological processes of general relevance [49]. In this regard, the initial discovery of the cause of HFTC unveiled ppGalNacT3 as a new element critical for maintaining Pi homeostasis [15]. Specifically, ppGalNacT3 catalyzes the initial steps of O-linked oligosaccharide biosynthesis, which consists in the transfer of an N-acetyl-D-galactosamine residue to a serine or threonine.

ppGalNacT3 deficiency has been shown to cause two distinct diseases, HFTC [815,42] and hyperostosis hyperphosphatemia syndrome [23,50,51], a recessive bone disorder. Thus, despite the fact that O-linked oligosaccharide biosynthesis occurs ubiquitously, ppGalNacT3 deficiency results in a very restricted spectrum of clinical manifestations. Functional redundancy among glycosyltransferases [52] has been invoked to explain this discrepancy [15]. However this is unlikely to account alone for the preferential accumulation of calcium deposits in the skin, suggesting a specific effect of ppGalNacT3 deficiency in this tissue. We therefore sought to assess the effects of GALNT3 down-regulation on dermal fibroblasts in an in vitro setting to address this issue in isolation from the systemic effects of ppGalNacT3. We show here that Pi induces ppGalNacT3 expression in dermal hTERT-fibroblasts and kidney epithelial cells. The fact that Pi modulates ppGalNacT3 expression in peripheral tissues may be interpreted to suggest that ppGalNacT3 plays a physiological role in peripheral tissues of relevance to the pathogenesis of HFTC. Accordingly, we used cultures of HFTC patients’ primary fibroblasts as well as human hTERT-fibroblasts transfected with GALNT3-specific siRNA to mimic the consequences of decreased ppGalNacT3 in an in vitro system. We discovered that decreased GALNT3 expression results in increased secretion of both FGF7 and MMPs, the expression of which is induced by FGF7. MMPs have been linked to ectopic calcification in vascular tissues. In particular, MMP-9 was found to be up-regulated during vascular calcification, and inhibition of MMPs activity was found to prevent calcification in the same model [30,31]. Thus, the present data together with previous observations suggest a causative role for MMPs in the pathogenesis of ectopic calcification in skin tissues. A number of studies indicate that Pi is able to induce MMPs expression in fibroblasts and other tissues [5356], suggesting that both ppGalNacT3 deficiency and induced MMP expression may have an additive detrimental (tissue-specific?) effect. This hypothesis is further supported by the finding that tetracyclines, which are known inhibitors of MMPs, have beneficial therapeutic effects in patients with cutaneous calcinosis [30,31,57,58]. This therapeutic response to tetracyclines is of particular importance as a large panoply of other treatments aimed at reducing circulating Pi levels shows very limited therapeutic benefit to HFTC patients [7,5961].

Collectively, the present observations imply that in addition to the well-known systemic effects of ppGalNacT3 deficiency on renal Pi re-absorption, the pathogenesis of HFTC may also involve local/peripheral processes. A number of natural modulators of ectopic calcification, such as fetuin-A, matrix Gla protein and osteopontin, may also play a local role in the pathogenesis of ectopic calcification in various tissues including the skin [6264]. The existence of such a wide array of biological molecules and pathways contributing to the formation of ectopic calcifications suggests that variant alleles encoding these proteins may account for the extensive clinical heterogeneity observed in HFTC and related disorders.

Acknowledgments

We wish to thank Sarah Selig for the gift of the Fibro-hTert cells. This study was supported in part by grants provided by Israel Science Foundation, the Rappaport Institute for Research in the Medical Sciences, Technion, Chief Scientists Office, Israel Ministry of Health and NIH/NIAMS grant R01 AR05262.

Appendix A. Supplementary data

Supplementary data associated with this article can be found, in the online version, at doi:10.1016/j.bbadis.2008.09.016.

References

  • 1.Renkema KY, Alexander RT, Bindels RJ, Hoenderop JG. Calcium and phosphate homeostasis: concerted interplay of new regulators. Ann Med. 2008;40:82–91. doi: 10.1080/07853890701689645. [DOI] [PubMed] [Google Scholar]
  • 2.Kestenbaum B. Phosphate metabolism in the setting of chronic kidney disease: significance and recommendations for treatment. Semin Dial. 2007;20:286–294. doi: 10.1111/j.1525-139X.2007.00303.x. [DOI] [PubMed] [Google Scholar]
  • 3.Melamed ML, Eustace JA, Plantinga L, Jaar BG, Fink NE, Coresh J, Klag MJ, Powe NR. Changes in serum calcium, phosphate, and PTH and the risk of death in incident dialysis patients: a longitudinal study. Kidney Int. 2006;70:351–357. doi: 10.1038/sj.ki.5001542. [DOI] [PubMed] [Google Scholar]
  • 4.Tonelli M, Sacks F, Pfeffer M, Gao Z, Curhan G. Relation between serum phosphate level and cardiovascular event rate in people with coronary disease. Circulation. 2005;112:2627–2633. doi: 10.1161/CIRCULATIONAHA.105.553198. [DOI] [PubMed] [Google Scholar]
  • 5.Caudarella R, Vescini F, Buffa A, Francucci CM. Hyperphosphatemia: effects on bone metabolism and cardiovascular risk. J Endocrinol Invest. 2007;30:29–34. [PubMed] [Google Scholar]
  • 6.White KE, Larsson TE, Econs MJ. The roles of specific genes implicated as circulating factors involved in normal and disordered phosphate homeostasis: frizzled related protein-4, matrix extracellular phosphoglycoprotein, and fibroblast growth factor 23. Endocrine Rev. 2006;27:221–241. doi: 10.1210/er.2005-0019. [DOI] [PubMed] [Google Scholar]
  • 7.Sprecher E. Tumoral calcinosis: new insights for the rheumatologist into a familial crystal deposition disease. Curr Rheumatol Re. 2007;9:237–242. doi: 10.1007/s11926-007-0038-6. [DOI] [PubMed] [Google Scholar]
  • 8.Garringer HJ, Mortazavi SM, Esteghamat F, Malekpour M, Boztepe H, Tanakol R, Davis SI, White KE. Two novel GALNT3 mutations in familial tumoral calcinosis. Am J Med Genet. 2007;143A:2390–2396. doi: 10.1002/ajmg.a.31947. [DOI] [PubMed] [Google Scholar]
  • 9.Barbieri AM, Filopanti M, Bua G, Beck-Peccoz P. Two novel nonsense mutations in GALNT3 gene are responsible for familial tumoral calcinosis. J Hum Gen. 2007;52:464–468. doi: 10.1007/s10038-007-0126-5. [DOI] [PubMed] [Google Scholar]
  • 10.Ichikawa S, Imel EA, Sorenson AH, Severe R, Knudson P, Harris GJ, Shaker JL, Econs MJ. Tumoral calcinosis presenting with eyelid calcifications due to novel missense mutations in the glycosyl transferase domain of the GALNT3 gene. J Clin Endocrinol Metabol. 2006;91:4472–4475. doi: 10.1210/jc.2006-1247. [DOI] [PubMed] [Google Scholar]
  • 11.Garringer HJ, Fisher C, Larsson TE, Davis SI, Koller DL, Cullen MJ, Draman MS, Conlon N, Jain A, Fedarko NS, Dasgupta B, White KE. The role of mutant UDP-N-acetyl-alpha-D-galactosamine-polypeptide N-acetylgalactosaminyltransferase 3 in regulating serum intact fibroblast growth factor 23 and matrix extracellular phosphoglycoprotein in heritable tumoral calcinosis. J Clin Endocrinol Metabol. 2006;91:4037–4042. doi: 10.1210/jc.2006-0305. [DOI] [PubMed] [Google Scholar]
  • 12.Campagnoli MF, Pucci A, Garelli E, Carando A, Defilippi C, Lala R, Ingrosso G, Dianzani I, Forni M, Ramenghi U. Familial tumoral calcinosis and testicular microlithiasis associated with a new mutation of GALNT3 in a white family. J Clin Pathol. 2006;59:440–442. doi: 10.1136/jcp.2005.026369. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Specktor P, Cooper JG, Indelman M, Sprecher E. Hyperphosphatemic familial tumoral calcinosis caused by a mutation in GALNT3 in a European kindred. J Hum Genet. 2006;51:487–490. doi: 10.1007/s10038-006-0377-6. [DOI] [PubMed] [Google Scholar]
  • 14.Ichikawa S, Lyles KW, Econs MJ. A novel GALNT3 mutation in a pseudoautosomal dominant form of tumoral calcinosis: evidence that the disorder is autosomal recessive. J Clin Endocrinol Metabol. 2005;90:2420–2423. doi: 10.1210/jc.2004-2302. [DOI] [PubMed] [Google Scholar]
  • 15.Topaz O, Shurman DL, Bergman R, Indelman M, Ratajczak P, Mizrachi M, Khamaysi Z, Behar D, Petronius D, Friedman V, Zelikovic I, Raimer S, Metzker A, Richard G, Sprecher E. Mutations in GALNT3, encoding a protein involved in O-linked glycosylation, cause familial tumoral calcinosis. Nat Genet. 2004;36:579–581. doi: 10.1038/ng1358. [DOI] [PubMed] [Google Scholar]
  • 16.Bennett EP, Hassan H, Clausen H. cDNA cloning and expression of a novel human UDP-N-acetyl-alpha-D-galactosamine. Polypeptide N-acetylgalactosaminyltransferase, GalNAc-t3. J Biol Chem. 1996;271:17006–17012. doi: 10.1074/jbc.271.29.17006. [DOI] [PubMed] [Google Scholar]
  • 17.Araya K, Fukumoto S, Backenroth R, Takeuchi Y, Nakayama K, Ito N, Yoshii N, Yamazaki Y, Yamashita T, Silver J, Igarashi T, Fujita T. A novel mutation in fibroblast growth factor 23 gene as a cause of tumoral calcinosis. J Clin Endocrinol Metabol. 2005;90:5523–5527. doi: 10.1210/jc.2005-0301. [DOI] [PubMed] [Google Scholar]
  • 18.Benet-Pages A, Orlik P, Strom TM, Lorenz-Depiereux B. An FGF23 missense mutation causes familial tumoral calcinosis with hyperphosphatemia. Hum Mol Genet. 2005;14:385–390. doi: 10.1093/hmg/ddi034. [DOI] [PubMed] [Google Scholar]
  • 19.Chefetz I, Heller R, Galli-Tsinopoulou A, Richard G, Wollnik B, Indelman M, Koerber F, Topaz O, Bergman R, Sprecher E, Schoenau E. A novel homozygous missense mutation in FGF23 causes Familial Tumoral Calcinosis associated with disseminated visceral calcification. Hum Genet. 2005;118:261–266. doi: 10.1007/s00439-005-0026-8. [DOI] [PubMed] [Google Scholar]
  • 20.Larsson T, Yu X, Davis SI, Draman MS, Mooney SD, Cullen MJ, White KE. A novel recessive mutation in fibroblast growth factor-23 causes familial tumoral calcinosis. J Clin Endocrinol Metabol. 2005;90:2424–2427. doi: 10.1210/jc.2004-2238. [DOI] [PubMed] [Google Scholar]
  • 21.Stubbs J, Liu S, Quarles LD. Role of fibroblast growth factor 23 in phosphate homeostasis and pathogenesis of disordered mineral metabolism in chronic kidney disease. Semin Dial. 2007;20:302–308. doi: 10.1111/j.1525-139X.2007.00308.x. [DOI] [PubMed] [Google Scholar]
  • 22.Yu X, White KE. FGF23 and disorders of phosphate homeostasis. Cytokine Growth Factor Rev. 2005;16:221–232. doi: 10.1016/j.cytogfr.2005.01.002. [DOI] [PubMed] [Google Scholar]
  • 23.Frishberg Y, Ito N, Rinat C, Yamazaki Y, Feinstein S, Urakawa I, Navon-Elkan P, Becker-Cohen R, Yamashita T, Araya K, Igarashi T, Fujita T, Fukumoto S. Hyperostosis-hyperphosphatemia syndrome: a congenital disorder of O-glycosylation associated with augmented processing of fibroblast growth factor 23. J Bone Miner Res. 2007;22:235–242. doi: 10.1359/jbmr.061105. [DOI] [PubMed] [Google Scholar]
  • 24.Kato K, Jeanneau C, Tarp MA, Benet-Pages A, Lorenz-Depiereux B, Bennett EP, Mandel U, Strom TM, Clausen H. Polypeptide GalNAc-transferase T3 and familial tumoral calcinosis. Secretion of fibroblast growth factor 23 requires O-glycosylation. J Biol Chem. 2006;281:18370–18377. doi: 10.1074/jbc.M602469200. [DOI] [PubMed] [Google Scholar]
  • 25.Larsson T, Davis SI, Garringer HJ, Mooney SD, Draman MS, Cullen MJ, White KE. Fibroblast growth factor-23 mutants causing familial tumoral calcinosis are differentially processed. Endocrinology. 2005;146:3883–3891. doi: 10.1210/en.2005-0431. [DOI] [PubMed] [Google Scholar]
  • 26.Ichikawa S, Imel EA, Kreiter ML, Yu X, Mackenzie DS, Sorenson AH, Goetz R, Mohammadi M, White KE, Econs MJ. A homozygous missense mutation in human KLOTHO causes severe tumoral calcinosis. J Muscul Neuron Inter. 2007;7:318–319. [PubMed] [Google Scholar]
  • 27.Ichikawa S, Imel EA, Kreiter ML, Yu X, Mackenzie DS, Sorenson AH, Goetz R, Mohammadi M, White KE, Econs MJ. A homozygous missense mutation in human KLOTHO causes severe tumoral calcinosis. J Clin Invest. 2007;117:2684–2691. doi: 10.1172/JCI31330. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Urakawa I, Yamazaki Y, Shimada T, Iijima K, Hasegawa H, Okawa K, Fujita T, Fukumoto S, Yamashita T. Klotho converts canonical FGF receptor into a specific receptor for FGF23. Nature. 2006;444:770–774. doi: 10.1038/nature05315. [DOI] [PubMed] [Google Scholar]
  • 29.Saito H, Kusano K, Kinosaki M, Ito H, Hirata M, Segawa H, Miyamoto K, Fukushima N. Human fibroblast growth factor-23 mutants suppress Na+-dependent phosphate co-transport activity and 1alpha,25-dihydroxyvitamin D3 production. J Biol Chem. 2003;278:2206–2211. doi: 10.1074/jbc.M207872200. [DOI] [PubMed] [Google Scholar]
  • 30.Bouvet C, Moreau S, Blanchette J, de Blois D, Moreau P. Sequential activation of matrix metalloproteinase 9 and transforming growth factor beta in arterial elastocalcinosis. Arterioscler Thromb Vasc Biol. 2008;28:856–862. doi: 10.1161/ATVBAHA.107.153056. [DOI] [PubMed] [Google Scholar]
  • 31.Qin X, Corriere MA, Matrisian LM, Guzman RJ. Matrix metalloproteinase inhibition attenuates aortic calcification. Arterioscler Thromb Vasc Biol. 2006;26:1510–1516. doi: 10.1161/01.ATV.0000225807.76419.a7. [DOI] [PubMed] [Google Scholar]
  • 32.Scatena M, Liaw L, Giachelli CM. Osteopontin: a multifunctional molecule regulating chronic inflammation and vascular disease. Arterioscler Thromb Vasc Biol. 2007;27:2302–2309. doi: 10.1161/ATVBAHA.107.144824. [DOI] [PubMed] [Google Scholar]
  • 33.Simionescu A, Simionescu DT, Vyavahare NR. Osteogenic responses in fibroblasts activated by elastin degradation products and transforming growth factor-beta1: role of myofibroblasts in vascular calcification. Am J Pathol. 2007;171:116–123. doi: 10.2353/ajpath.2007.060930. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Bodnar AG, Ouellette M, Frolkis M, Holt SE, Chiu CP, Morin GB, Harley CB, Shay JW, Lichtsteiner S, Wright WE. Extension of life-span by introduction of telomerase into normal human cells. Science. 1998;279:349–352. doi: 10.1126/science.279.5349.349. [DOI] [PubMed] [Google Scholar]
  • 35.Nomoto M, Izumi H, Ise T, Kato K, Takano H, Nagatani G, Shibao K, Ohta R, Imamura T, Kuwano M, Matsuo K, Yamada Y, Itoh H, Kohno K. Structural basis for the regulation of UDP-N-acetyl-alpha-D-galactosamine: polypeptide N-acetylgalactosaminyl transferase-3 gene expression in adenocarcinoma cells. Cancer Res. 1999;59:6214–6222. [PubMed] [Google Scholar]
  • 36.Dosaka-Akita H, Kinoshita I, Yamazaki K, Izumi H, Itoh T, Katoh H, Nishimura M, Matsuo K, Yamada Y, Kohno K. N-acetylgalactosaminyl transferase-3 is a potential new marker for non-small cell lung cancers. Br J Cancer. 2002;87:751–755. doi: 10.1038/sj.bjc.6600536. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.White AR, Du T, Laughton KM, Volitakis I, Sharples RA, Xilinas ME, Hoke DE, Holsinger RM, Evin G, Cherny RA, Hill AF, Barnham KJ, Li QX, Bush AI, Masters CL. Degradation of the Alzheimer disease amyloid beta-peptide by metal-dependent up-regulation of metalloprotease activity. J Biol Chem. 2006;281:17670–17680. doi: 10.1074/jbc.M602487200. [DOI] [PubMed] [Google Scholar]
  • 38.Meleti Z, Shapiro IM, Adams CS. Inorganic phosphate induces apoptosis of osteoblast-like cells in culture. Bone. 2000;27:359–366. doi: 10.1016/s8756-3282(00)00346-x. [DOI] [PubMed] [Google Scholar]
  • 39.Szczepanska-Konkel M, Yusufi AN, VanScoy M, Webster SK, Dousa TP. Phosphonocarboxylic acids as specific inhibitors of Na+-dependent transport of phosphate across renal brush border membrane. J Biol Chem. 1986;261:6375–6383. [PubMed] [Google Scholar]
  • 40.Carpenter TO, Ellis BK, Insogna KL, Philbrick WM, Sterpka J, Shimkets R. Fibroblast growth factor 7: an inhibitor of phosphate transport derived from oncogenic osteomalacia-causing tumors. J Clin Endocrinol Metabol. 2005;90:1012–1020. doi: 10.1210/jc.2004-0357. [DOI] [PubMed] [Google Scholar]
  • 41.auf demKeller U, Krampert M, Kumin A, Braun S, Werner S. Keratinocyte growth factor: effects on keratinocytes and mechanisms of action. Eur J Cell Biol. 2004;83:607–612. doi: 10.1078/0171-9335-00389. [DOI] [PubMed] [Google Scholar]
  • 42.Topaz O, Bergman R, Mandel U, Maor G, Goldberg R, Richard G, Sprecher E. Absence of intraepidermal glycosyltransferase ppGalNac-T3 expression in familial tumoral calcinosis. Am J Dermatopathol. 2005;27:211–215. doi: 10.1097/01.dad.0000158298.02545.a5. [DOI] [PubMed] [Google Scholar]
  • 43.Madlener M, Mauch C, Conca W, Brauchle M, Parks WC, Werner S. Regulation of the expression of stromelysin-2 by growth factors in keratinocytes: implications for normal and impaired wound healing. Biochem J. 1996;320(Pt 2):659–664. doi: 10.1042/bj3200659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ropiquet F, Huguenin S, Villette JM, Ronfie V, Le Brun G, Maitland NJ, Cussenot O, Fiet J, Berthon P. FGF7/KGF triggers cell transformation and invasion on immortalised human prostatic epithelial PNT1A cells. Int J Cancer. 1999;82:237–243. doi: 10.1002/(sici)1097-0215(19990719)82:2<237::aid-ijc14>3.0.co;2-q. [DOI] [PubMed] [Google Scholar]
  • 45.Shin EY, Ma EK, Kim CK, Kwak SJ, Kim EG. Src/ERK but not phospholipase D is involved in keratinocyte growth factor-stimulated secretion of matrix metallo-protease-9 and urokinase-type plasminogen activator in SNU-16 human stomach cancer cell. J Cancer Res Clin Oncol. 2002;128:596–602. doi: 10.1007/s00432-002-0388-4. [DOI] [PubMed] [Google Scholar]
  • 46.Simian M, Hirai Y, Navre M, Werb Z, Lochter A, Bissell MJ. The interplay of matrix metalloproteinases, morphogens and growth factors is necessary for branching of mammary epithelial cells. Development. 2001;128:3117–3131. doi: 10.1242/dev.128.16.3117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Fukumoto S. Physiological regulation and disorders of phosphate metabolism–pivotal role of fibroblast growth factor 23. Intern Med. 2008;47:337–343. doi: 10.2169/internalmedicine.47.0730. [DOI] [PubMed] [Google Scholar]
  • 48.Uribarri J. Phosphorus homeostasis in normal health and in chronic kidney disease patients with special emphasis on dietary phosphorus intake. Semin Dial. 2007;20:295–301. doi: 10.1111/j.1525-139X.2007.00309.x. [DOI] [PubMed] [Google Scholar]
  • 49.Antonarakis SE, Beckmann JS. Mendelian disorders deserve more attention. Nat Rev Genet. 2006;7:277–282. doi: 10.1038/nrg1826. [DOI] [PubMed] [Google Scholar]
  • 50.Olauson H, Krajisnik T, Larsson C, Lindberg B, Larsson TE. A novel missense mutation in GALNT3 causing hyperostosis-hyperphosphataemia syndrome. Eur J Endocrinol. 2008;158:929–934. doi: 10.1530/EJE-08-0011. [DOI] [PubMed] [Google Scholar]
  • 51.Ichikawa S, Guigonis V, Imel EA, Courouble M, Heissat S, Henley JD, Sorenson AH, Petit B, Lienhardt A, Econs MJ. Novel GALNT3 mutations causing hyperostosis-hyperphosphatemia syndrome result in low intact fibroblast growth factor 23 concentrations. J Clin Endocrinol Metabol. 2007;92:1943–1947. doi: 10.1210/jc.2006-1825. [DOI] [PubMed] [Google Scholar]
  • 52.Ten Hagen KG, Fritz TA, Tabak LA. All in the family: the UDP-GalNAc: polypeptide N-acetylgalactosaminyltransferases. Glycobiology. 2003;13:1R–16R. doi: 10.1093/glycob/cwg007. [DOI] [PubMed] [Google Scholar]
  • 53.Molloy ES, Morgan MP, Doherty GA, McDonnell B, O’Byrne J, Fitzgerald DJ, McCarthy GM. Mechanism of basic calcium phosphate crystal-stimulated matrix metalloproteinase-13 expression in osteoarthritic synovial fibroblasts: inhibition by prostaglandin E2. Ann Rheum Dis. 2008 doi: 10.1136/ard.2007.079582. in press. [DOI] [PubMed] [Google Scholar]
  • 54.Reuben PM, Wenger L, Cruz M, Cheung HS. Induction of matrix metalloproteinase-8 in human fibroblasts by basic calcium phosphate and calcium pyrophosphate dihydrate crystals: effect of phosphocitrate. Connect Tissue Res. 2001;42:1–12. doi: 10.3109/03008200109014244. [DOI] [PubMed] [Google Scholar]
  • 55.Bai G, Howell DS, Howard GA, Roos BA, Cheung HS. Basic calcium phosphate crystals up-regulate metalloproteinases but down-regulate tissue inhibitor of metalloproteinase-1 and -2 in human fibroblasts. Osteoarthritis Cartilage. 2001;9:416–422. doi: 10.1053/joca.2000.0407. [DOI] [PubMed] [Google Scholar]
  • 56.McCarthy GM, Macius AM, Christopherson PA, Ryan LM, Pourmotabbed T. Basic calcium phosphate crystals induce synthesis and secretion of 92 kDa gelatinase (gelatinase B/matrix metalloprotease 9) in human fibroblasts. Ann Rheum Dis. 1998;57:56–60. doi: 10.1136/ard.57.1.56. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Boulman N, Slobodin G, Rozenbaum M, Rosner I. Calcinosis in rheumatic diseases. Semin Arthritis Rheum. 2005;34:805–812. doi: 10.1016/j.semarthrit.2005.01.016. [DOI] [PubMed] [Google Scholar]
  • 58.Cohen H, Solomon V, Alferiev IS, Breuer E, Ornoy A, Patlas N, Eidelman N, Hagele G, Golomb G. Bisphosphonates and tetracycline: experimental models for their evaluation in calcium-related disorders. Pharm Res. 1998;15:606–613. doi: 10.1023/a:1011990129437. [DOI] [PubMed] [Google Scholar]
  • 59.Slavin RE, Wen J, Kumar D, Evans EB. Familial tumoral calcinosis. A clinical, histopathologic, and ultrastructural study with an analysis of its calcifying process and pathogenesis. Am J Surg Pathol. 1993;17:788–802. [PubMed] [Google Scholar]
  • 60.Steinherz R, Chesney RW, Eisenstein B, Metzker A, DeLuca HF, Phelps M. Elevated serum calcitriol concentrations do not fall in response to hyperphosphatemia in familial tumoral calcinosis. Am J Dis Child. 1960;139(1985):816–819. doi: 10.1001/archpedi.1985.02140100078036. [DOI] [PubMed] [Google Scholar]
  • 61.Metzker A, Eisenstein B, Oren J, Samuel R. Tumoral calcinosis revisited–common and uncommon features. Report of ten cases and review. Eur J Ped. 1988;147:128–132. doi: 10.1007/BF00442209. [DOI] [PubMed] [Google Scholar]
  • 62.Hendig D, Schulz V, Arndt M, Szliska C, Kleesiek K, Gotting C. Role of serum fetuin-A, a major inhibitor of systemic calcification, in pseudoxanthoma elasticum. Clin Chem. 2006;52:227–234. doi: 10.1373/clinchem.2005.059253. [DOI] [PubMed] [Google Scholar]
  • 63.Hendig D, Arndt M, Szliska C, Kleesiek K, Gotting C. SPP1 promoter polymorphisms: identification of the first modifier gene for pseudoxanthoma elasticum. Clin Chem. 2007;53:829–836. doi: 10.1373/clinchem.2006.083675. [DOI] [PubMed] [Google Scholar]
  • 64.Jiang Q, Li Q, Uitto J. Aberrant mineralization of connective tissues in a mouse model of pseudoxanthoma elasticum: systemic and local regulatory factors. J Invest Dermatol. 2007;127:1392–1402. doi: 10.1038/sj.jid.5700729. [DOI] [PubMed] [Google Scholar]

RESOURCES